151
|
Exum NG, Lee GO, Olórtegui MP, Yori PP, Salas MS, Trigoso DR, Colston JM, Schwab KJ, McCormick BJJ, Kosek MN. A Longitudinal Study of Household Water, Sanitation, and Hygiene Characteristics and Environmental Enteropathy Markers in Children Less than 24 Months in Iquitos, Peru. Am J Trop Med Hyg 2018; 98:995-1004. [PMID: 29436350 PMCID: PMC5928816 DOI: 10.4269/ajtmh.17-0464] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Poor child gut health, resulting from a lack of access to an improved toilet or clean water, has been proposed as a biological mechanism underlying child stunting and oral vaccine failure. Characteristics related to household sanitation, water use, and hygiene were measured among a birth cohort of 270 children from peri-urban Iquitos Peru. These children had monthly stool samples and urine samples at four time points and serum samples at (2–4) time points analyzed for biomarkers related to intestinal inflammation and permeability. We found that less storage of fecal matter near the household along with a reliable water connection were associated with reduced inflammation, most prominently the fecal biomarker myeloperoxidase (MPO) (no sanitation facility compared with those with an onsite toilet had −0.43 log MPO, 95% confidence interval [CI]: −0.74, −0.13; and households with an intermittent connection versus those with a continuous supply had +0.36 log MPO, 95% CI: 0.08, 0.63). These results provide preliminary evidence for the hypothesis that children less than 24 months of age living in unsanitary conditions will have elevated gut inflammation.
Collapse
Affiliation(s)
- Natalie G Exum
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Gwenyth O Lee
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Maribel Paredes Olórtegui
- Asociación Benéfica Proyectos de Informática, Salud, Medicina, y Agricultura (A.B. PRISMA), Iquitos, Peru.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Pablo Peñataro Yori
- Asociación Benéfica Proyectos de Informática, Salud, Medicina, y Agricultura (A.B. PRISMA), Iquitos, Peru.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Mery Siguas Salas
- Asociación Benéfica Proyectos de Informática, Salud, Medicina, y Agricultura (A.B. PRISMA), Iquitos, Peru
| | - Dixner Rengifo Trigoso
- Asociación Benéfica Proyectos de Informática, Salud, Medicina, y Agricultura (A.B. PRISMA), Iquitos, Peru
| | - Josh M Colston
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Kellogg J Schwab
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | | | - Margaret N Kosek
- Asociación Benéfica Proyectos de Informática, Salud, Medicina, y Agricultura (A.B. PRISMA), Iquitos, Peru.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
152
|
François R, Yori PP, Rouhani S, Siguas Salas M, Paredes Olortegui M, Rengifo Trigoso D, Pisanic N, Burga R, Meza R, Meza Sanchez G, Gregory MJ, Houpt ER, Platts-Mills JA, Kosek MN. The other Campylobacters: Not innocent bystanders in endemic diarrhea and dysentery in children in low-income settings. PLoS Negl Trop Dis 2018; 12:e0006200. [PMID: 29415075 PMCID: PMC5819825 DOI: 10.1371/journal.pntd.0006200] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 02/20/2018] [Accepted: 12/31/2017] [Indexed: 12/13/2022] Open
Abstract
Background Campylobacter is one of the main causes of gastroenteritis worldwide. Most of the current knowledge about the epidemiology of this food-borne infection concerns two species, C. coli and C. jejuni. Recent studies conducted in developing countries and using novel diagnostic techniques have generated evidence of the increasing burden and importance of other Campylobacter species, i.e. non-C. coli/jejuni. We performed a nested case-control study to compare the prevalence of C. coli/jejuni and other Campylobacter in children with clinical dysentery and severe diarrhea as well as without diarrhea to better understand the clinical importance of infections with Campylobacter species other than C. coli/jejuni. Methodology/Principal findings Our nested case-control study of 439 stool samples included dysenteric stools, stools collected during severe diarrhea episodes, and asymptomatic stools which were systematically selected to be representative of clinical phenotypes from 9,160 stools collected during a birth cohort study of 201 children followed until two years of age. Other Campylobacter accounted for 76.4% of the 216 Campylobacter detections by qPCR and were more prevalent than C. coli/jejuni across all clinical groups. Other Campylobacter were also more prevalent than C. coli/jejuni across all age groups, with older children bearing a higher burden of other Campylobacter. Biomarkers of intestinal inflammation and injury (methylene blue, fecal occult test, myeloperoxidase or MPO) showed a strong association with dysentery, but mixed results with infection. MPO levels were generally higher among children infected with C. coli/jejuni, but Shigella-infected children suffering from dysentery recorded the highest levels (26,224 ng/mL); the lowest levels (10,625 ng/mL) were among asymptomatic children infected with other Campylobacter. Adjusting for age, sex, and Shigella infection, dysentery was significantly associated with C. coli/jejuni but not with other Campylobacter, whereas severe diarrhea was significantly associated with both C. coli/jejuni and other Campylobacter. Compared to asymptomatic children, children suffering from dysentery had a 14.6 odds of C. coli/jejuni infection (p-value < 0.001, 95% CI 5.5–38.7) but were equally likely to have other Campylobacter infections–odds ratio of 1.3 (0.434, 0.7–2.4). Children suffering from severe diarrhea were more likely than asymptomatic children to test positive for both C. coli/jejuni and other Campylobacter–OR of 2.8 (0.034, 1.1–7.1) and 1.9 (0.018, 1.1–3.1), respectively. Compared to the Campylobacter-free group, the odds of all diarrhea given C. coli/jejuni infection and other Campylobacter infection were 8.8 (<0.001, 3.0–25.7) and 2.4 (0.002, 1.4–4.2), respectively. Eliminating other Campylobacter in this population would eliminate 24.9% of the diarrhea cases, which is almost twice the population attributable fraction of 15.1% due to C. coli/jejuni. Conclusions/Significance Eighty-seven percent of the dysentery and 59.5% of the severe diarrhea samples were positive for Campylobacter, Shigella, or both, emphasizing the importance of targeting these pathogens to limit the impact of dysentery and severe diarrhea in children. Notably, the higher prevalence of other Campylobacter compared to C. coli/jejuni, their increasing burden during early childhood, and their association with severe diarrhea highlight the importance of these non-C. coli/jejuni Campylobacter species and suggest a need to clarify their importance in the etiology of clinical disease across different epidemiological contexts. Campylobacter is a major public health concern in developed and developing countries. C. coli and C. jejuni have long been considered to be the major disease-causing species, and clinical microbiologic approaches target these two species. However, less selective diagnostic approaches have shown the increasing importance of other Campylobacter species (i.e. non-C. coli/jejuni). Our case-control study investigated the association between diarrhea, C. coli/jejuni, and other Campylobacter among 439 stool samples from 201 children in peri-urban communities in Loreto, Peru. Three quarters of the 216 Campylobacter detections were associated with other Campylobacter, whose prevalence increased with age and was greater than that of C. coli/jejuni in all age and clinical groups (dysentery, severe diarrhea, and asymptomatic). Despite their lower prevalence, C. coli/jejuni were more strongly associated with higher levels of myeloperoxidase, clinical dysentery, and the presence of leukocytes and blood in the stool compared to other Campylobacter. Other Campylobacter were equally likely as C. coli/jejuni to be detected in severe diarrhea cases–odds ratio of 1.9 (p-value = 0.018, 95% CI 1.1–3.1) and 2.8 (0.034, 1.1–7.1), respectively. Removing C. coli/jejuni in this population would eliminate 15.1% of diarrhea compared to 24.9% if other Campylobacter were eliminated.
Collapse
Affiliation(s)
- Ruthly François
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
| | - Pablo Peñataro Yori
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
- Biomedical Research, Asociación Benéfica PRISMA, Iquitos, Peru
| | - Saba Rouhani
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
| | | | | | | | - Nora Pisanic
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
| | - Rosa Burga
- Bacteriology Department, Naval Medical Research Unit-6 (NAMRU-6), Lima, Peru
| | - Rina Meza
- Bacteriology Department, Naval Medical Research Unit-6 (NAMRU-6), Lima, Peru
| | - Graciela Meza Sanchez
- Facultad de Medicina Humana, Universidad Nacional de la Amazonía Peruana, Iquitos, Peru
| | - Michael J. Gregory
- Bacteriology Department, Naval Medical Research Unit-6 (NAMRU-6), Lima, Peru
| | - Eric R. Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, United States of America
| | - James A. Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, United States of America
| | - Margaret N. Kosek
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
- Biomedical Research, Asociación Benéfica PRISMA, Iquitos, Peru
- Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, United States of America
- * E-mail:
| |
Collapse
|
153
|
Enteroaggregative Escherichia coli Subclinical Infection and Coinfections and Impaired Child Growth in the MAL-ED Cohort Study. J Pediatr Gastroenterol Nutr 2018; 66:325-333. [PMID: 29356769 DOI: 10.1097/mpg.0000000000001717] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE We evaluated the impact of subclinical enteroaggregative Escherichia coli (EAEC) infection alone and in combination with other pathogens in the first 6 months of life on child growth. METHODS Nondiarrheal samples from 1684 children across 8 Multisite Birth Cohort Study, Malnutrition and Enteric Diseases (MAL-ED) sites in Asia, Africa, and Latin America were tested monthly; more than 90% of children were followed-up twice weekly for the first 6 months of life. RESULTS Children with subclinical EAEC infection did not show altered growth between enrollment and 6 months. Conversely, EAEC coinfection with any other pathogen was negatively associated with delta weight-for-length (P < 0.05) and weight-for-age (P > 0.05) z scores between 0 and 6 months. The presence of 2 or more pathogens without EAEC was not significantly associated with delta weight-for-length and weight-for-age. The most frequent EAEC coinfections included Campylobacter spp, heat-labile toxin-producing enterotoxigenic E coli, Cryptosporidium spp, and atypical enteropathogenic E coli. Myeloperoxidase levels were increased with EAEC coinfection (P < 0.05). EAEC pathogen codetection was associated with lower neopterin levels compared to those of no-pathogen control children (P < 0.05). Mothers of children with EAEC coinfections had lower levels of education, poorer hygiene and sanitation, lower socioeconomic status, and lower breast-feeding rates compared to mothers of children in whom no pathogen was detected (P < 0.05). CONCLUSIONS These data emphasize the public health importance of subclinical EAEC infection in early infancy in association with other pathogens and the need for improved maternal and child care, hygiene, sanitation, and socioeconomic factors.
Collapse
|
154
|
Shores DR, Everett AD. Children as Biomarker Orphans: Progress in the Field of Pediatric Biomarkers. J Pediatr 2018; 193:14-20.e31. [PMID: 29031860 PMCID: PMC5794519 DOI: 10.1016/j.jpeds.2017.08.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/04/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Darla R Shores
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Allen D Everett
- Division of Cardiology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
155
|
Harper KM, Mutasa M, Prendergast AJ, Humphrey J, Manges AR. Environmental enteric dysfunction pathways and child stunting: A systematic review. PLoS Negl Trop Dis 2018; 12:e0006205. [PMID: 29351288 PMCID: PMC5792022 DOI: 10.1371/journal.pntd.0006205] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/31/2018] [Accepted: 01/03/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is commonly defined as an acquired subclinical disorder of the small intestine, characterized by villous atrophy and crypt hyperplasia. EED has been proposed to underlie stunted growth among children in developing countries. A collection of biomarkers, organized into distinct domains, has been used to measure different aspects of EED. Here, we examine whether these hypothesized relationships, among EED domains and between each domain and stunting, are supported by data from recent studies. METHODOLOGY A systematic literature search was conducted using PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL between January 1, 2010 and April 20, 2017. Information on study objective, design, population, location, biomarkers, and results were recorded, as well as qualitative and quantitative definitions of EED. Biomarkers were organized into five EED domains, and the number of studies that support or do not support relationships among domains and between each domain with stunting were summarized. RESULTS There was little evidence to support the pathway from intestinal permeability to microbial translocation and from microbial translocation to stunting, but stronger support existed for the link between intestinal inflammation and systemic inflammation and for intestinal inflammation and stunting. There was conflicting evidence for the pathways from intestinal damage to intestinal permeability and intestinal damage to stunting. CONCLUSIONS These results suggest that certain EED biomarkers may require reconsideration, particularly those most difficult to measure, such as microbial translocation and intestinal permeability. We discuss several issues with currently used biomarkers and recommend further analysis of pathogen-induced changes to the intestinal microbiota as a pathway leading to stunting.
Collapse
Affiliation(s)
- Kaitlyn M. Harper
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Maxine Mutasa
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jean Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Amee R. Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- British Columbia Centre for Disease Control, Vancouver, Canada
| |
Collapse
|
156
|
Mwape I, Bosomprah S, Mwaba J, Mwila-Kazimbaya K, Laban NM, Chisenga CC, Sijumbila G, Simuyandi M, Chilengi R. Immunogenicity of rotavirus vaccine (RotarixTM) in infants with environmental enteric dysfunction. PLoS One 2017; 12:e0187761. [PMID: 29281659 PMCID: PMC5744930 DOI: 10.1371/journal.pone.0187761] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction Deployment of rotavirus vaccines has contributed to significant declines in diarrheal morbidity and mortality globally. Unfortunately, vaccine performance in low-middle income countries (LMICs) is generally lower than in developed countries. The cause for this has been associated with several host and maternal factors including poor water sanitation and hygiene (WASH) status, which are predominant in LMICs. More recently, environmental enteric dysfunction (EED) has specifically been hypothesized to contribute to poor vaccine uptake and response. The aim of this study was to examine the association between serological biomarkers of EED and seroconversion to rotavirus vaccine in Zambian infants. Methods This was a retrospective cohort study of 142 infants who had been fully immunized with Rotarix™, and had known seroconversion status. Seroconversion was defined as 4-fold or more increase in rotavirus-specific IgA titres between pre-vaccination and one month post-dose two vaccination. We performed ELISA assays to assess soluble CD14 (sCD14), Endotoxin Core IgG Antibodies (EndoCAb), intestinal fatty acid binding protein (i-FABP) and Zonulin according to the manufacturers protocols. Generalised linear model with family-poisson, link-log and robust standard error was used to estimate the independent effects of biomarkers on seroconversion adjusting for important cofounders. Results The median concentration of Zonulin, Soluble CD14, EndoCaB, and IFABP were 209.3 (IQR = 39.7, 395.1), 21.5 (IQR = 21.5, 21.5), 0.3 (IQR = 0.3, 0.3), and 107.7 (IQR = 6.4, 1141.4) respectively. In multivariable analyses adjusting for the independent effect of other biomarkers and confounders (i.e. age of child at vaccination, breast-milk anti-rotavirus IgA, infant serum anti-rotavirus IgG, and IgA seropositivity at baseline), there was strong evidence of about 24% increase in seroconversion due to doubling Zonulin concentration (Adjusted risk ratio (aRR) = 1.24; 95% CI = 1.12 to1.37; p<0.0001). Similarly, we found about 7% increase in seroconversion due to doubling IFABP concentration (aRR = 1.07; 95% CI = 1.02 to 1.13; p = 0.006). Conclusion We found that high levels of zonulin and IFABP played a role in seroconversion. It is plausible that increased gut permeability in EED allows greater uptake of the live virus within the vaccine, but later consequences result in deleterious local structural distortions and malabsorption syndromes.
Collapse
Affiliation(s)
- Innocent Mwape
- Center for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of Physiological sciences,University of Zambia, Lusaka, Zambia
- * E-mail:
| | - Samuel Bosomprah
- Center for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of Biostatistics, School of Public Health, University of Ghana, Legon, Accra, Ghana
| | - John Mwaba
- Center for Infectious Disease Research in Zambia, Lusaka, Zambia
| | | | | | | | - Gibson Sijumbila
- Department of Physiological sciences,University of Zambia, Lusaka, Zambia
| | | | - Roma Chilengi
- Center for Infectious Disease Research in Zambia, Lusaka, Zambia
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
157
|
George CM, Burrowes V, Perin J, Oldja L, Biswas S, Sack D, Ahmed S, Haque R, Bhuiyan NA, Parvin T, Bhuyian SI, Akter M, Li S, Natarajan G, Shahnaij M, Faruque AG, Stine OC. Enteric Infections in Young Children are Associated with Environmental Enteropathy and Impaired Growth. Trop Med Int Health 2017; 23:26-33. [PMID: 29121442 DOI: 10.1111/tmi.13002] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the relationship between faecal contamination in child play spaces, enteric infections, environmental enteropathy (EE) and impaired growth among young children. METHODS A prospective cohort study was conducted of 203 children 6-30 months of age in rural Bangladesh. Stool samples were analysed by quantitative PCR for Shigella, Enterotoxigenic Escherichia coli (ETEC), Campylobacter jejuni, Giardia intestinalis and Cryptosporidium spp. Four faecal markers of intestinal inflammation were also measured: alpha-1-antitrypsin, myeloperoxidase, neopterin and calprotectin. Child growth was measured at baseline and 9 months after enrolment. E. coli was measured in soil in child play spaces. RESULTS Forty-seven percent of study children had three or more enteric pathogens in their stool. Thirty five percent (71/203) of children had Shigella, 30% (61/203) had ETEC, 73% (148/203) had C. jejuni, 79% (160/203) had Giardia intestinalis and none had Cryptosporidium. Children with ETEC had significantly higher calprotectin concentrations (Coefficient: 1.35, 95% Confidence Interval [CI]: 1.005, 1.82). Children with Shigella had a significantly higher odds of being stunted at our 9-month follow-up (OR: 2.01, 95% CI: 1.02, 3.93). Children with Giardia intestinalis had significantly higher E.coli counts in the soil collected from their play spaces (OR: 1.23, 95% CI: 1.02, 1.48). CONCLUSION Enteric infections were significantly associated with EE and impaired growth in rural Bangladesh. These findings provide further evidence to support the hypothesis that contaminated soil in child play spaces can lead to enteric infections, many of which are likely subclinical, resulting in EE and impaired growth in young children.
Collapse
Affiliation(s)
- Christine Marie George
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vanessa Burrowes
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jamie Perin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lauren Oldja
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shwapon Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.,Department of Internal Medicine, Rangpur Medical College Hospital, Rangpur, Bangladesh
| | - David Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shahnawaz Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Nurul Amin Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Mahmuda Akter
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shan Li
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gayathri Natarajan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mohammad Shahnaij
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Abu G Faruque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - O Colin Stine
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
158
|
Nataro JP, Guerrant RL. Chronic consequences on human health induced by microbial pathogens: Growth faltering among children in developing countries. Vaccine 2017; 35:6807-6812. [DOI: 10.1016/j.vaccine.2017.05.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/28/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
|
159
|
Abstract
The global impact of childhood malnutrition is staggering. The synergism between malnutrition and infection contributes substantially to childhood morbidity and mortality. Anthropometric indicators of malnutrition are associated with the increased risk and severity of infections caused by many pathogens, including viruses, bacteria, protozoa, and helminths. Since childhood malnutrition commonly involves the inadequate intake of protein and calories, with superimposed micronutrient deficiencies, the causal factors involved in impaired host defense are usually not defined. This review focuses on literature related to impaired host defense and the risk of infection in primary childhood malnutrition. Particular attention is given to longitudinal and prospective cohort human studies and studies of experimental animal models that address causal, mechanistic relationships between malnutrition and host defense. Protein and micronutrient deficiencies impact the hematopoietic and lymphoid organs and compromise both innate and adaptive immune functions. Malnutrition-related changes in intestinal microbiota contribute to growth faltering and dysregulated inflammation and immune function. Although substantial progress has been made in understanding the malnutrition-infection synergism, critical gaps in our understanding remain. We highlight the need for mechanistic studies that can lead to targeted interventions to improve host defense and reduce the morbidity and mortality of infectious diseases in this vulnerable population.
Collapse
|
160
|
Abstract
Malnutrition contributes significantly to death and illness worldwide and especially to the deaths of children younger than 5 years. The relation between intestinal changes in malnutrition and morbidity and mortality has not been well characterized; however, recent research indicates that the functional and morphologic changes of the intestine secondary to malnutrition itself contribute significantly to these negative clinical outcomes and may be potent targets of intervention. The aim of this review was to summarize current knowledge of experimental and clinically observed changes in the intestine from malnutrition preclinical models and human studies. Limited clinical studies have shown villous blunting, intestinal inflammation, and changes in the intestinal microbiome of malnourished children. In addition to these findings, experimental data using various animal models of malnutrition have found evidence of increased intestinal permeability, upregulated intestinal inflammation, and loss of goblet cells. More mechanistic studies are urgently needed to improve our understanding of malnutrition-related intestinal dysfunction and to identify potential novel targets for intervention.
Collapse
|
161
|
Duh J, Spears D. Health and Hunger: Disease, Energy Needs, and the Indian Calorie Consumption Puzzle. ECONOMIC JOURNAL (LONDON, ENGLAND) 2017; 127:2378-2409. [PMID: 33612850 PMCID: PMC7797625 DOI: 10.1111/ecoj.12417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 05/08/2019] [Accepted: 08/13/2019] [Indexed: 05/22/2023]
Abstract
India's experience presents a puzzle at odds with a basic fact of household economics: amidst unprecedented economic growth, average per capita daily calorie consumption has declined in recent decades. Does an improving disease environment explain the calorie decline? A diminished burden of infectious disease could lower energy needs by increasing absorption and effective use of calories. We document a robust effect of disease exposure - measured as infant mortality and as poor sanitation - on calorie consumption. Similar effects are found using multiple datasets and empirical strategies. Disease can account for an important fraction (one-fifth or more) of India's calorie decline.
Collapse
Affiliation(s)
- Josephine Duh
- University of Texas at Austin, Indian Statistical Institute - Delhi Centre, and r.i.c.e
| | - Dean Spears
- University of Texas at Austin, Indian Statistical Institute - Delhi Centre, and r.i.c.e
| |
Collapse
|
162
|
Pernica JM, Steenhoff AP, Mokomane M, Moorad B, Lechiile K, Smieja M, Mazhani L, Cheng J, Kelly MS, Loeb M, Stordal K, Goldfarb DM. Rapid enteric testing to permit targeted antimicrobial therapy, with and without Lactobacillus reuteri probiotics, for paediatric acute diarrhoeal disease in Botswana: A pilot, randomized, factorial, controlled trial. PLoS One 2017; 12:e0185177. [PMID: 28991918 PMCID: PMC5633142 DOI: 10.1371/journal.pone.0185177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/03/2017] [Indexed: 01/29/2023] Open
Abstract
Introduction Diarrhoeal disease is the second-leading cause of death in young children. Current guidelines recommend treating children with acute non-bloody diarrhea with oral rehydration solutions and zinc, but not antimicrobials. However, in many resource-limited settings, infections with treatable enteric bacterial and protozoan pathogens are common. Probiotics have shown promise as an adjunct treatment for diarrhoea but have not been studied in sub-Saharan Africa. Methods We conducted a pilot, factorial, randomized, placebo-controlled trial of children aged 2–60 months hospitalized in Botswana for acute non-bloody diarrhoea. A rapid test-and-treat intervention, consisting of multiplex PCR testing of rectal swabs taken at enrolment, accompanied by targeted antimicrobial therapy if treatable pathogens were detected, was compared to the reference standard of no stool testing. Additionally, Lactobacillus reuteri DSM 17938 x 60 days was compared to placebo treatment. The main objective of this pilot study was to assess feasibility. The primary clinical outcome was the increase in age-standardized height (HAZ) at 60 days adjusted for baseline HAZ. Results Seventy-six patients were enrolled over a seven-month study period. We judged that the recruitment rate, lab processing times, communication protocols, provision of specific antimicrobials, and follow-up rates were acceptable. Compared to the reference arm (no stool testing and placebo treatment), the combination of the rapid test-and-treat strategy plus L. reuteri DSM 17938 was associated with an increase of 0.61 HAZ (95% CI 0.09–1.13) and 93% lower odds of recurrent diarrhoea (OR 0.07, 95%CI 0.01–0.61) at 60 days. Discussion We demonstrated that it was feasible to evaluate the study interventions in Botswana. Despite the small sample size, we observed a statistically significant increase in HAZ at 60 days and significantly lower odds of recurrent diarrhoea in children receiving both rapid test-and-treat and L. reuteri. There is sufficient evidence to warrant proceeding with a larger follow-up trial in a similar setting.
Collapse
Affiliation(s)
- Jeffrey M. Pernica
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| | - Andrew P. Steenhoff
- Center for Global Health & Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Botswana-UPenn Partnership, Gaborone, Botswana
| | - Margaret Mokomane
- Botswana Ministry of Health, National Health Laboratory, Gaborone, Botswana
| | | | | | - Marek Smieja
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Loeto Mazhani
- Department of Paediatrics and Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Ji Cheng
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S. Kelly
- Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Mark Loeb
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - David M. Goldfarb
- Department of Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
163
|
Abstract
PURPOSE OF REVIEW Giardia is a common intestinal parasite worldwide, and infection can be associated with clear and sometimes persistent symptomatology. However, in children in high-prevalence settings, it is not associated with or is perhaps even protective against acute diarrhea, and the association with long-term outcomes has been difficult to discern. RECENT FINDINGS Recent studies have made progress in helping us disentangle this apparent paradox. First, prospective, well-characterized cohort studies have added to the data on the association between Giardia and diarrhea in these settings and have further characterized associations between Giardia infection and nutrition, gut function, and growth. Second, animal models have further characterized the host response to Giardia and helped elucidate mechanisms by which Giardia could impair child development. Finally, new work has shed light on the heterogeneity of human Giardia strains, which may both explain discrepant findings in the literature and help guide higher-resolution analyses of this pathogen in the future. SUMMARY The true clinical impact of endemic pediatric giardiasis remains unclear, but recent prospective studies have confirmed a high prevalence of persistent, subclinical Giardia infections and associated growth shortfalls. Integrating how nutritional, microbial, metabolic, and pathogen-strain variables influence these outcomes could sharpen delineations between pathogenic and potentially beneficial attributes of this enigmatic parasite.
Collapse
|
164
|
Denno DM, Tarr PI, Nataro JP. Environmental Enteric Dysfunction: A Case Definition for Intervention Trials. Am J Trop Med Hyg 2017; 97:1643-1646. [PMID: 29016294 PMCID: PMC5805039 DOI: 10.4269/ajtmh.17-0183] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Donna M Denno
- Department of Pediatrics, University of Washington, Seattle, Washington.,Department of Global Health, University of Washington, Seattle, Washington
| | - Phillip I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
165
|
Dorshow RB, Hall-Moore C, Shaikh N, Talcott MR, Faubion WA, Rogers TE, Shieh JJ, Debreczeny MP, Johnson JR, Dyer RB, Singh RJ, Tarr PI. Measurement of gut permeability using fluorescent tracer agent technology. Sci Rep 2017; 7:10888. [PMID: 28883476 PMCID: PMC5589723 DOI: 10.1038/s41598-017-09971-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
The healthy gut restricts macromolecular and bacterial movement across tight junctions, while increased intestinal permeability accompanies many intestinal disorders. Dual sugar absorption tests, which measure intestinal permeability in humans, present challenges. Therefore, we asked if enterally administered fluorescent tracers could ascertain mucosal integrity, because transcutaneous measurement of differentially absorbed molecules could enable specimen-free evaluation of permeability. We induced small bowel injury in rats using high- (15 mg/kg), intermediate- (10 mg/kg), and low- (5 mg/kg) dose indomethacin. Then, we compared urinary ratios of enterally administered fluorescent tracers MB-402 and MB-301 to urinary ratios of sugar tracers lactulose and rhamnose. We also tested the ability of transcutaneous sensors to measure the ratios of absorbed fluorophores. Urinary fluorophore and sugar ratios reflect gut injury in an indomethacin dose dependent manner. The fluorophores generated smooth curvilinear ratio trajectories with wide dynamic ranges. The more chaotic sugar ratios had narrower dynamic ranges. Fluorophore ratios measured through the skin distinguished indomethacin-challenged from same day control rats. Enterally administered fluorophores can identify intestinal injury in a rat model. Fluorophore ratios are measureable through the skin, obviating drawbacks of dual sugar absorption tests. Pending validation, this technology should be considered for human use.
Collapse
Affiliation(s)
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michael R Talcott
- Division of Comparative Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - William A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Roy B Dyer
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, MN, USA
| | | | - Phillip I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
166
|
Ngobeni R, Abhyankar MM, Jiang NM, Farr LA, Samie A, Haque R, Moonah SN. Entamoeba histolytica-Encoded Homolog of Macrophage Migration Inhibitory Factor Contributes to Mucosal Inflammation during Amebic Colitis. J Infect Dis 2017; 215:1294-1302. [PMID: 28186296 DOI: 10.1093/infdis/jix076] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
Understanding the mechanisms by which Entamoeba histolytica drives gut inflammation is critical for the development of improved preventive and therapeutic strategies. E. histolytica encodes a homolog of the human cytokine macrophage migration inhibitory factor (MIF). Here, we investigated the role of E. histolytica MIF (EhMIF) during infection. We found that the concentration of fecal EhMIF correlated with the level of intestinal inflammation in persons with intestinal amebiasis. Mice treated with antibodies that specifically block EhMIF had reduced chemokine expression and neutrophil infiltration in the mucosa. In addition to antibody-mediated neutralization, we used a genetic approach to test the effect of EhMIF on mucosal inflammation. Mice infected with parasites overexpressing EhMIF had increased chemokine expression, neutrophil influx, and mucosal damage. Together, these results uncover a specific parasite protein that increases mucosal inflammation, expands our knowledge of host-parasite interaction during amebic colitis, and highlights a potential immunomodulatory target.
Collapse
Affiliation(s)
- Renay Ngobeni
- Department of Medicine, University of Virginia Health System, Charlottesville
| | | | - Nona M Jiang
- Department of Medicine, University of Virginia Health System, Charlottesville
| | - Laura A Farr
- Department of Medicine, University of Virginia Health System, Charlottesville
| | - Amidou Samie
- Department of Microbiology, University of Venda, Limpopo Province, South Africa; and
| | - Rashidul Haque
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Shannon N Moonah
- Department of Medicine, University of Virginia Health System, Charlottesville
| |
Collapse
|
167
|
Abstract
Environmental enteropathy is a chronic condition of the small intestine associated with increased intestinal permeability, mucosal inflammation, malabsorption, and systemic inflammation. It is commonly accompanied by enteric infections and is misleadingly considered a subclinical disease. Potential effects of enteric infections and enteropathy on vaccine responses, child growth, cognitive development, and even later life obesity, diabetes, and metabolic syndrome are increasingly being recognized. Herein, we review the evolving challenges to defining environmental enteropathy and enteric infections, current evidence for the magnitude and determinants of its burden, new assessment tools, and relevant interventions.
Collapse
Affiliation(s)
- Elizabeth T Rogawski
- Department of Public Health Sciences, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA; Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA.
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA
| |
Collapse
|
168
|
Wit JM, Himes JH, van Buuren S, Denno DM, Suchdev PS. Practical Application of Linear Growth Measurements in Clinical Research in Low- and Middle-Income Countries. Horm Res Paediatr 2017; 88:79-90. [PMID: 28196362 PMCID: PMC5804842 DOI: 10.1159/000456007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/10/2017] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND/AIMS Childhood stunting is a prevalent problem in low- and middle-income countries and is associated with long-term adverse neurodevelopment and health outcomes. In this review, we define indicators of growth, discuss key challenges in their analysis and application, and offer suggestions for indicator selection in clinical research contexts. METHODS Critical review of the literature. RESULTS Linear growth is commonly expressed as length-for-age or height-for-age z-score (HAZ) in comparison to normative growth standards. Conditional HAZ corrects for regression to the mean where growth changes relate to previous status. In longitudinal studies, growth can be expressed as ΔHAZ at 2 time points. Multilevel modeling is preferable when more measurements per individual child are available over time. Height velocity z-score reference standards are available for children under the age of 2 years. Adjusting for covariates or confounders (e.g., birth weight, gestational age, sex, parental height, maternal education, socioeconomic status) is recommended in growth analyses. CONCLUSION The most suitable indicator(s) for linear growth can be selected based on the number of available measurements per child and the child's age. By following a step-by-step algorithm, growth analyses can be precisely and accurately performed to allow for improved comparability within and between studies.
Collapse
Affiliation(s)
- Jan M. Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands,*Jan M. Wit, MD, PhD, Department of Pediatrics, J6S, Leiden University Medical Center, PO Box 9600, NL-2300RC Leiden (The Netherlands), E-Mail
| | - John H. Himes
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stef van Buuren
- Netherlands Organization for Applied Scientific Research TNO, Leiden, the Netherlands,Department of Methodology and Statistics, Faculty of Social and Behavioral Sciences, University of Utrecht, Utrecht, the Netherlands
| | - Donna M. Denno
- Department of Pediatrics and Global Health, University of Washington, Seattle, Washington, USA
| | - Parminder S. Suchdev
- Department of Pediatrics and Hubert Department of Global Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
169
|
Abstract
OBJECTIVES Environmental enteric dysfunction (EED) may inhibit growth and development in low- and middle-income countries, but available assessment methodologies limit its study. In rural Bangladesh, we measured EED using the widely used lactulose mannitol ratio (L:M) test and a panel of intestinal and systemic health biomarkers to evaluate convergence among biomarkers and describe risk factors for EED. METHODS In 539 18-month-old children finishing participation in a randomized food supplementation trial, serum, stool, and urine collected after lactulose and mannitol dosing were analyzed for biomarkers of intestinal absorption, inflammation, permeability and repair, and systemic inflammation. EED scores for each participant were developed using principal component analysis and partial least squares regression. Associations between scores and L:M and with child sociodemographic and health characteristics were evaluated using regression analysis. RESULTS EED prevalence (L:M > 0.07) was 39.0%; 60% had elevated acute phase proteins (C-reactive protein >5 mg/L or α-1 acid glycoprotein >100 mg/dL). Correlations between intestinal biomarkers were low, with the highest between myeloperoxidase and α-1 antitrypsin (r = 0.33, P < 0.01), and biomarker values did not differ by supplementation history. A 1-factor partial least squares model with L:M as the dependent variable explained only 8.6% of L:M variability. In adjusted models, L:M was associated with child sex and socioeconomic status index, whereas systemic inflammation was predicted mainly by recent illness, not EED. CONCLUSIONS Impaired intestinal health is widespread in this setting of prevalent stunting, but a panel of serum and stool biomarkers demonstrated poor agreement with L:M. Etiologies of intestinal and systemic inflammation are likely numerous and complex in resource-poor settings, underscoring the need for a better case definition with corresponding diagnostic methods to further the study of EED.
Collapse
|
170
|
Jimenez L, Duggan CP. Biomarkers of Environmental Enteric Dysfunction: The Good, the Bad, and the Ugly. J Pediatr Gastroenterol Nutr 2017; 65:4-5. [PMID: 28644342 PMCID: PMC5492894 DOI: 10.1097/mpg.0000000000001591] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Lissette Jimenez
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | | |
Collapse
|
171
|
Lee GO, McCormick BJJ, Seidman JC, Kosek MN, Haque R, Olortegui MP, Lima AAM, Bhutta ZA, Kang G, Samie A, Amour C, Mason CJ, Ahmed T, Yori PP, Oliveira DB, Alam D, Babji S, Bessong P, Mduma E, Shrestha SK, Ambikapathi R, Lang DR, Gottlieb M, Guerrant RL, Caulfield LE. Infant Nutritional Status, Feeding Practices, Enteropathogen Exposure, Socioeconomic Status, and Illness Are Associated with Gut Barrier Function As Assessed by the Lactulose Mannitol Test in the MAL-ED Birth Cohort. Am J Trop Med Hyg 2017; 97:281-290. [PMID: 28719336 PMCID: PMC5508897 DOI: 10.4269/ajtmh.16-0830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 01/31/2017] [Indexed: 12/20/2022] Open
Abstract
The lactulose mannitol (LM) dual sugar permeability test is the most commonly used test of environmental enteropathy in developing countries. However, there is a large but conflicting literature on its association with enteric infection and host nutritional status. We conducted a longitudinal cohort using a single field protocol and comparable laboratory procedures to examine intestinal permeability in multiple, geographically diverse pediatric populations. Using a previously published systematic review to guide the selection of factors potentially associated with LM test results, we examined the relationships between these factors and mucosal breach, represented by percent lactulose excretion; absorptive area, represented by percent mannitol excretion; and gut barrier function, represented by the L/M ratio. A total of 6,602 LM tests were conducted in 1,980 children at 3, 6, 9, and 15 months old; percent lactulose excretion, percent mannitol excretion, and the L/M ratio were expressed as age- and sex-specific normalized values using the Brazil cohort as the reference population. Among the factors considered, recent severe diarrhea, lower socioeconomic status, and recent asymptomatic enteropathogen infections were associated with decreased percent mannitol excretion and higher L/M ratios. Poorer concurrent weight-for-age, infection, and recent breastfeeding were associated with increased percent lactulose excretion and increased L/M ratios. Our results support previously reported associations between the L/M ratio and factors related to child nutritional status and enteropathogen exposure. These results were remarkably consistent across sites and support the hypothesis that the frequency of these exposures in communities living in poverty leads to alterations in gut barrier function.
Collapse
Affiliation(s)
- Gwenyth O. Lee
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | | | - Jessica C. Seidman
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
| | - Margaret N. Kosek
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | | | - Aldo A. M. Lima
- Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Zulfiqar A. Bhutta
- Center of Excellence in Women and Child Health, the Aga Khan University, Karachi, Pakistan
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Amidou Samie
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | | | - Carl J. Mason
- Walter Reed/Armed Forces Research Institute of Medical Sciences, Kathmandu, Nepal
| | | | - Pablo Peñataro Yori
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Didar Alam
- Center of Excellence in Women and Child Health, the Aga Khan University, Karachi, Pakistan
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Pascal Bessong
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | | | - Sanjaya K. Shrestha
- Walter Reed/Armed Forces Research Institute of Medical Sciences, Kathmandu, Nepal
| | - Ramya Ambikapathi
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Dennis R. Lang
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Foundation for the NIH, Bethesda, Maryland
| | | | - Richard L. Guerrant
- Division of Infectious Diseases, University of Virginia, Charlottesville, Virginia
| | - Laura E. Caulfield
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | |
Collapse
|
172
|
Colston JM, Peñataro Yori P, Colantuoni E, Moulton LH, Ambikapathi R, Lee G, Rengifo Trigoso D, Siguas Salas M, Kosek MN. A methodologic framework for modeling and assessing biomarkers of environmental enteropathy as predictors of growth in infants: an example from a Peruvian birth cohort. Am J Clin Nutr 2017; 106:245-255. [PMID: 28592604 DOI: 10.3945/ajcn.116.151886] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/09/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Environmental enteropathy (EE) impairs the gut's absorptive capacity and immune function and causes decelerations in statural growth that manifest gradually over time.Objective: To illustrate an approach for assessing emerging biomarkers of EE, we separately assessed the associations between 3 such markers and subsequent nutritional status.Design: Stool samples were routinely collected between January 2010 and November 2014 from a cohort of 303 Peruvian infants and analyzed for concentrations of the biomarkers α-1-antitrypsin (AAT), myeloperoxidase, and neopterin. For each marker, a mixed-effects linear regression model was fitted for length-for-age z scores (LAZs) obtained from anthropometric assessments that incorporated covariate predictors, polynomial terms for age, and product interaction terms to test associations over varying lag lengths. The biomarkers' contribution to the models was assessed with the use of the likelihood ratio test and partial R2 statistics.Results: Test statistics for the combined inclusion of the 4-model terms that involved the biomarker were highly statistically significant for AAT (28.71; P < 0.0001) and myeloperoxidase (62.79; P < 0.0001) over a 3-mo lag and moderately so for neopterin (13.97; P = 0.0074). AAT and myeloperoxidase seemed to interact strongly with age, with the magnitude and direction of the effect varying considerably over the first 3 y of life. The largest proportion of the variance explained by any biomarker (2.8%) and the largest difference in LAZ predicted between the 5th and 95th percentile (0.25) was by myeloperoxidase over a 2-mo lag.Conclusions: Of the 3 fecal biomarkers studied, 2 that related to intestinal function-AAT and myeloperoxidase-were associated with small but highly statistically significant differences in future statural growth trajectories in infants in this cohort, lending further evidence to the EE hypothesis that increased gut permeability and inflammation adversely affects subsequent nutritional status. This association exhibited a complex interaction with age. This trial was registered at clinicaltrials.gov as NCT02441426.
Collapse
Affiliation(s)
| | | | | | - Lawrence H Moulton
- Departments of International Health and.,Biostatistics, Johns Hopkins School of Public Health, Baltimore, MD
| | | | - Gwenyth Lee
- Department of Epidemiology, University of Michigan, Ann Arbor, MI; and
| | - Dixner Rengifo Trigoso
- Biomedical Research Unit, Asociación Benéfica PRISMA (PRISMA Charitable Association), Iquitos, Peru
| | - Mery Siguas Salas
- Biomedical Research Unit, Asociación Benéfica PRISMA (PRISMA Charitable Association), Iquitos, Peru
| | | |
Collapse
|
173
|
Garzón M, Pereira-da-Silva L, Seixas J, Papoila AL, Alves M, Ferreira F, Reis A. Association of enteric parasitic infections with intestinal inflammation and permeability in asymptomatic infants of São Tomé Island. Pathog Glob Health 2017; 111:116-127. [PMID: 28279129 PMCID: PMC5445637 DOI: 10.1080/20477724.2017.1299831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The cumulative effect of repeated asymptomatic enteric infections on intestinal barrier is not fully understood in infants. We aimed to evaluate the association between previous enteric parasitic infections and intestinal inflammation and permeability at 24-months of age, in asymptomatic infants of São Tomé Island. A subset of infants from a birth cohort, with intestinal parasite evaluations in at least four points of assessment, was eligible. Intestinal inflammatory response and permeability were assessed using fecal S100A12 and alpha-1-antitrypsin (A1AT), respectively. The cutoff <-1SD for weight-for-length and length-for-age was used to define wasting and stunting. Multivariable linear regression analysis explored if cumulative enteric parasitic infections explained variability of fecal biomarkers, after adjusting for potential confounders. Eighty infants were included. Giardia duodenalis and soil-transmitted helminths (STH) were the most frequent parasites. The median (interquartile range) levels were 2.87 μg/g (2.41-3.92) for S100A12 and 165.1 μg/g (66.0-275.6) for A1AT. Weak evidence of association was found between S100A12 levels and G. duodenalis (p = 0.080) and STH infections (p = 0.089), and between A1AT levels and parasitic infection of any etiology (p = 0.089), at 24-months of age. Significant associations between A1AT levels and wasting (p = 0.006) and stunting (p = 0.044) were found. Previous parasitic infections were not associated with fecal biomarkers at 24 months of age. To summarize, previous asymptomatic parasitic infections showed no association with intestinal barrier dysfunction. Notwithstanding, a tendency toward increased levels of the inflammatory biomarker was observed for current G. duodenalis and STH infections, and increased levels of the permeability biomarker were significantly associated with stunting and wasting.
Collapse
Affiliation(s)
- Marisol Garzón
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Luis Pereira-da-Silva
- Research Unit, Centro Hospitalar de Lisboa Central, Lisboa, Portugal
- Woman, Children and Adolescent’s Medicine Teaching and Research Area, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jorge Seixas
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ana Luísa Papoila
- Research Unit, Centro Hospitalar de Lisboa Central, Lisboa, Portugal
- NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Marta Alves
- Research Unit, Centro Hospitalar de Lisboa Central, Lisboa, Portugal
| | - Filipa Ferreira
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ana Reis
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
174
|
Morita T, Perin J, Oldja L, Biswas S, Sack RB, Ahmed S, Haque R, Bhuiyan NA, Parvin T, Bhuyian SI, Akter M, Talukder KA, Shahnaij M, Faruque AG, George CM. Mouthing of Soil Contaminated Objects is Associated with Environmental Enteropathy in Young Children. Trop Med Int Health 2017; 22:670-678. [PMID: 28319300 DOI: 10.1111/tmi.12869] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To characterise childhood mouthing behaviours and to investigate the association between object-to-mouth and food-to-mouth contacts, diarrhoea prevalence and environmental enteropathy. METHODS A prospective cohort study was conducted of 216 children ≤30 months of age in rural Bangladesh. Mouthing contacts with soil and food and objects with visible soil were assessed by 5-h structured observation. Stool was analysed for four faecal markers of intestinal inflammation: alpha-1-antitrypsin, myeloperoxidase, neopterin and calprotectin. RESULTS Overall 82% of children were observed mouthing soil, objects with visible soil, or food with visible soil during the structured observation period. Sixty two percent of children were observed mouthing objects with visible soil, 63% were observed mouthing food with visible soil, and 18% were observed mouthing soil only. Children observed mouthing objects with visible soil had significantly elevated faecal calprotectin concentrations (206.81 μg/g, 95% confidence interval [CI]: 6.27, 407.36). There was also a marginally significant association between Escherichia coli counts in soil from a child's play space and the prevalence rate of diarrhoea (diarrhoea prevalence ratio: 2.03, 95% CI 0.97, 4.25). CONCLUSION These findings provide further evidence to support the hypothesis that childhood mouthing behaviour in environments with faecal contamination can lead to environmental enteropathy in susceptible paediatric populations. Furthermore, these findings suggest that young children mouthing objects with soil, which occurred more frequently than soil directly (60% vs. 18%), was an important exposure route to faecal pathogens and a risk factor for environmental enteropathy.
Collapse
Affiliation(s)
- Tomohiko Morita
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jamie Perin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lauren Oldja
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shwapon Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.,Department of Internal Medicine, Rangpur Medical College Hospital, Rangpur, Bangladesh
| | - R Bradley Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shahnawaz Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Nurul Amin Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Mahmuda Akter
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Kaisar A Talukder
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mohammad Shahnaij
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Abu G Faruque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Christine Marie George
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
175
|
Kaur M, Graham JP, Eisenberg JNS. Livestock Ownership Among Rural Households and Child Morbidity and Mortality: An Analysis of Demographic Health Survey Data from 30 Sub-Saharan African Countries (2005-2015). Am J Trop Med Hyg 2017; 96:741-748. [PMID: 28044044 DOI: 10.4269/ajtmh.16-0664] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Children living in homes with livestock may have both an increased risk of enteric infections and improved access to food, and therefore improved nutritional status. Few studies, however, have characterized these relationships in tandem. This study investigated the association between child health and household ownership of livestock. A cross-sectional study was performed using data from Demographic and Health Surveys conducted in 30 sub-Saharan African countries with 215,971 rural children under 5 years of age from 2005 to 2015. Logistic regression was performed for each country to estimate the relationship between a log2 increase in the number of livestock owned by the household and three child-health outcomes: 2-week prevalence of diarrhea, stunting, and all-cause mortality. Results for each country were combined using meta-analyses. Most countries (22 of 30) displayed an odds ratio (OR) less than 1 for child stunting associated with livestock (pooled OR = 0.97; 95% confidence interval [CI] = 0.95, 0.99). The results for diarrhea were more even with 14 countries displaying ORs greater than 1 and 10 displaying ORs less than 1. Most countries (22 of 30) displayed an OR greater than 1 for child mortality (pooled OR = 1.04; 95% CI = 1.02, 1.06). All meta-analyses displayed significant heterogeneity by country. Our analysis is consistent with the theory that livestock may have a dual role as protective against stunting, an indicator of chronic malnutrition, and a risk factor for all-cause mortality in children, which may be linked to acute infections. The heterogeneity by country, however, indicates more data are needed on specific household livestock management practices.
Collapse
Affiliation(s)
- Maneet Kaur
- Division of Epidemiology, School of Public Health, University of California, Berkeley, California
| | | | - Joseph N S Eisenberg
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
176
|
The Association Between Fecal Biomarkers of Environmental Enteropathy and Rotavirus Vaccine Response in Nicaraguan Infants. Pediatr Infect Dis J 2017; 36:412-416. [PMID: 27977553 DOI: 10.1097/inf.0000000000001457] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Environmental enteropathy (EE) is a common intestinal condition among children living in low- and middle-income countries and is associated with diminished enteric immunity to gastrointestinal pathogens, and possibly to oral vaccine antigens. The goal of this study was to examine associations between biomarkers of EE and immunogenicity to the pentavalent rotavirus vaccine (RV5). METHODS Infants were recruited 1 day before their first RV5 immunization in León, Nicaragua, from public health rosters. Infants provided a preimmunization blood and stool sample, and a second blood sample 1 month after receipt of RV5. We measured immunoglobin A (IgA) seroconversion to the first dose of RV5 and concentrations of 4 previously identified fecal biomarkers of EE (alpha-1 antitrypsin, neopterin, myeloperoxidase and calprotectin). We then assessed associations between concentrations of these biomarkers, both individually and as combined scores, and seroconversion to the first dose of RV5. RESULTS Of the 43 enrolled infants, 24 (56%) seroconverted after the first dose of RV5. As compared with infants who seroconverted, those who did not seroconvert had higher median concentrations of both myeloperoxidase (3.1 vs. 1.1 µg/mL, P = 0.002) and calprotectin (199.1 vs. 156.2 µg/mL, P = 0.03). Further, those who did not seroconvert had a higher median combined score of the 4 biomarkers as compared with those who seroconverted (6.5 vs. 4.5, P = 0.017). CONCLUSIONS We found an association between biomarkers of EE and seroconversion to the first dose of RV5. It is possible that interventions that prevent or ameliorate EE may also improve oral rotavirus vaccine response.
Collapse
|
177
|
Bhutta ZA, Guerrant RL, Nelson CA. Neurodevelopment, Nutrition, and Inflammation: The Evolving Global Child Health Landscape. Pediatrics 2017; 139:S12-S22. [PMID: 28562245 DOI: 10.1542/peds.2016-2828d] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2016] [Indexed: 11/24/2022] Open
Abstract
The last decade has witnessed major reductions in child mortality and a focus on saving lives with key interventions targeting major causes of child deaths, such as neonatal deaths and those due to childhood diarrhea and pneumonia. With the transition to Sustainable Development Goals, the global health community is expanding child health initiatives to address not only the ongoing need for reduced mortality, but also to decrease morbidity and adverse exposures toward improving health and developmental outcomes. The relationship between adverse environmental exposures frequently associated with factors operating in the prepregnancy period and during fetal development is well established. Also well appreciated are the developmental impacts (both short- and long-term) associated with postnatal factors, such as immunostimulation and environmental enteropathy, and the additional risks posed by the confluence of factors related to malnutrition, poor living conditions, and the high burden of infections. This article provides our current thinking on the pathogenesis and risk factors for adverse developmental outcomes among young children, setting the scene for potential interventions that can ameliorate these adversities among families and children at risk.
Collapse
Affiliation(s)
- Zulfiqar A Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada; .,Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Charles A Nelson
- Laboratories of Cognitive Neuroscience, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; and.,Human Development Program, Harvard Graduate School of Education, Cambridge, Massachusetts
| |
Collapse
|
178
|
Krebs NF, Lozoff B, Georgieff MK. Neurodevelopment: The Impact of Nutrition and Inflammation During Infancy in Low-Resource Settings. Pediatrics 2017; 139:S50-S58. [PMID: 28562248 DOI: 10.1542/peds.2016-2828g] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2016] [Indexed: 11/24/2022] Open
Abstract
Infancy and early childhood (ie, birth through age 24 months) represent a period of life with both exquisite opportunity and vulnerability for neurodevelopment. This is due to rapid brain development, both anatomic and functional, as well as to high nutrient requirements during a time of dependence on human milk and complementary foods. Complex interactions exist among nutrition, social, and physical environments and exposures. The newborn brain also reflects maternal exposures that occurred as the product of many interacting forces during gestation. Connections between nutrient use and acute and chronic inflammation are increasingly recognized, but the evidence base linking both nutrition and inflammation to neurodevelopment is relatively modest and quite limited for this young age group specifically. This article provides an overview of key interactions of nutritional requirements relevant to brain development and function; nutritional vulnerabilities related to maternal nutritional status and function; and the impact of environmental exposures and inflammation on nutrient homeostasis and neurodevelopment during this critical developmental window.
Collapse
Affiliation(s)
- Nancy F Krebs
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado;
| | - Betsy Lozoff
- Center for Human Growth and Development and.,Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan; and
| | - Michael K Georgieff
- Division of Neonatology, University of Minnesota School of Medicine, Minneapolis, Minnesota
| |
Collapse
|
179
|
Suchdev PS, Boivin MJ, Forsyth BW, Georgieff MK, Guerrant RL, Nelson CA. Assessment of Neurodevelopment, Nutrition, and Inflammation From Fetal Life to Adolescence in Low-Resource Settings. Pediatrics 2017; 139:S23-S37. [PMID: 28562246 DOI: 10.1542/peds.2016-2828e] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2016] [Indexed: 11/24/2022] Open
Abstract
Efforts to improve child neurodevelopment are critical to health, equity, and sustainable development, particularly in low-resource settings in the United States and globally. The colliding epidemics of food insecurity, infectious diseases, and noncommunicable diseases interact and impact neurodevelopment. Understanding the complex relationships between nutrition, inflammation, and neurodevelopment can inform clinical and public health interventions to improve outcomes. This article reviews key definitions, tools, and considerations for the assessment of nutrition, inflammation, and child neurodevelopment. The effectiveness of existing assessment tools to reflect status and biology, particularly in relation to each other, and to predict long-term changes in health is examined. The aim of this review is to present the extant evidence, identify critical research gaps, and suggest a research agenda for future longitudinal and intervention studies to address the assessment of nutrition, inflammation, and child neurodevelopment, particularly in low-resource settings. Despite research gaps, there is a strong relationship between nutrition, inflammation, environmental factors, and child neurodevelopment, which emphasizes the need to evaluate targeted, early interventions to improve long-term health and well-being.
Collapse
Affiliation(s)
- Parminder S Suchdev
- Departments of Pediatrics and .,Global Health, Emory University, Atlanta, Georgia
| | - Michael J Boivin
- Departments of Psychiatry and.,Neurology and Ophthalmology, Michigan State University, East Lansing, Michigan
| | - Brian W Forsyth
- Department of Pediatrics, Yale University, New Haven, Connecticut
| | - Michael K Georgieff
- Departments of Pediatrics and.,Child Psychology, University of Minnesota School of Medicine, Minneapolis, Minnesota
| | - Richard L Guerrant
- Center for Global Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Charles A Nelson
- Laboratories of Cognitive Neuroscience, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston Massachusetts; and.,Harvard Graduate School of Education, Cambridge, Massachusetts
| |
Collapse
|
180
|
Reese H, Routray P, Torondel B, Sclar G, Delea MG, Sinharoy SS, Zambrano L, Caruso B, Mishra SR, Chang HH, Clasen T. Design and rationale of a matched cohort study to assess the effectiveness of a combined household-level piped water and sanitation intervention in rural Odisha, India. BMJ Open 2017; 7:e012719. [PMID: 28363920 PMCID: PMC5387990 DOI: 10.1136/bmjopen-2016-012719] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Government efforts to address massive shortfalls in rural water and sanitation in India have centred on construction of community water sources and toilets for selected households. However, deficiencies with water quality and quantity at the household level and community coverage and actual use of toilets have led Gram Vikas, a local non-governmental organization in Odisha, India, to develop an approach that provides household-level piped water connections contingent on full community-level toilet coverage. METHODS This matched cohort study was designed to assess the effectiveness of a combined piped water and sanitation intervention. Households with children <5 years in 45 randomly selected intervention villages and 45 matched control villages will be followed over 17 months. The primary outcome is prevalence of diarrhoeal diseases; secondary health outcomes include soil-transmitted helminth infection, nutritional status, seroconversion to enteric pathogens, urogenital infections and environmental enteric dysfunction. In addition, intervention effects on sanitation and water coverage, access and use, environmental fecal contamination, women's empowerment, as well as collective efficacy, and intervention cost and cost-effectiveness will be assessed. ETHICS AND DISSEMINATION The study protocol has been reviewed and approved by the ethics boards of the London School of Hygiene and Tropical Medicine, UK and KIIT University, Bhubaneswar, India. Findings will be disseminated via peer-reviewed literature and presentation to stakeholders, government officials, implementers and researchers. TRIAL REGISTRATION NUMBER NCT02441699.
Collapse
Affiliation(s)
- Heather Reese
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | | | - Belen Torondel
- London School of Hygiene and Tropical Medicine, London, UK
| | - Gloria Sclar
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Maryann G Delea
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- London School of Hygiene and Tropical Medicine, London, UK
| | - Sheela S Sinharoy
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Laura Zambrano
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Bethany Caruso
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Samir R Mishra
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Howard H Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Thomas Clasen
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
181
|
Sindhu KNC, Cunliffe N, Peak M, Turner M, Darby A, Grassly N, Gordon M, Dube Q, Babji S, Praharaj I, Verghese V, Iturriza-Gómara M, Kang G. Impact of maternal antibodies and infant gut microbiota on the immunogenicity of rotavirus vaccines in African, Indian and European infants: protocol for a prospective cohort study. BMJ Open 2017; 7:e016577. [PMID: 28360258 PMCID: PMC5372070 DOI: 10.1136/bmjopen-2017-016577] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Gastroenteritis is the leading cause of morbidity and mortality among young children living in resource-poor settings, majority of which is attributed to rotavirus. Rotavirus vaccination can therefore have a significant impact on infant mortality. However, rotavirus vaccine efficacy in Sub-Saharan Africa and Southeast Asia is significantly lower than in high-income countries. Maternally derived antibodies, infant gut microbiota and concomitant oral polio vaccination have been proposed as potential reasons for poor vaccine performance in low-income settings. The overall aim of this study is to compare the role of maternally derived antibodies and infant gut microbiota in determining immune response to rotavirus vaccine in high-income and low-income settings, using the same vaccine and a similar study protocol. METHODS AND ANALYSIS The study is an observational cohort in three countries-Malawi, India and UK. Mothers will be enrolled in third trimester of pregnancy and followed up, along with infants after delivery, until the infant completes two doses of oral rotavirus vaccine (along with routine immunisation). The levels of prevaccination maternally derived rotavirus-specific antibodies (IgG) will be correlated with infant seroconversion and antibody titres, 4 weeks after the second dose of rotavirus vaccine. Both within-country and between-country comparisons of gut microbiome will be carried out between children who seroconvert and those who do not. The impact of oral polio vaccine coadministration on rotavirus vaccine response will be studied in Indian infants. ETHICS AND DISSEMINATION Ethical approvals have been obtained from Integrated Research Application System (IRAS, NHS ethics) in UK, College of Medicine Research and Ethics Committee (COMREC) in Malawi and Institutional Review Board (IRB), Christian Medical College, Vellore in India. Participant recruitment and follow-up is ongoing at all three sites. Analysis of data, followed by publication of the results, is expected in 2018.
Collapse
Affiliation(s)
| | | | - Matthew Peak
- Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | | | | | | | | | | | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Ira Praharaj
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Valsan Verghese
- Department of Child Health, Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
182
|
Kosek MN. Causal Pathways from Enteropathogens to Environmental Enteropathy: Findings from the MAL-ED Birth Cohort Study. EBioMedicine 2017; 18:109-117. [PMID: 28396264 PMCID: PMC5405169 DOI: 10.1016/j.ebiom.2017.02.024] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 02/09/2017] [Accepted: 02/23/2017] [Indexed: 01/23/2023] Open
Abstract
Children living in these settings had a high prevalence of enteropathogens, high levels of intestinal inflammation, abnormal intestinal permeability, high markers of systemic inflammation, and postnatal acquired linear growth deficits when compared to children living in the US or Europe This study contributes empiric evidence to demonstrate that enteric infection alters both fecal markers of inflammation and permeability Current markers of enteropathy fail to account for a large portion of the observed shortfalls in linear growth in these populations, and markers of systemic inflammation appear as the most promising predictive biomarkers for identifying linear growth failure in children
Environmental enteropathy (EE) is hypothesized as a mediator of growth faltering, but few prospective studies have evaluated pathways linking enteropathogen exposure, intestinal inflammation and permeability, and growth. The MAL-ED study represents a novel analytical framework and explicitly evaluates multiple putative EE pathways in combination and using an unprecedented quantity of data. Despite evidence that gut inflammation and altered gut permeability are frequently present and that associations between enteropathogen exposure and gut dysfunction exist, the observed attributable effects of EE on growth faltering in young children were small. Background Environmental enteropathy (EE), the adverse impact of frequent and numerous enteric infections on the gut resulting in a state of persistent immune activation and altered permeability, has been proposed as a key determinant of growth failure in children in low- and middle-income populations. A theory-driven systems model to critically evaluate pathways through which enteropathogens, gut permeability, and intestinal and systemic inflammation affect child growth was conducted within the framework of the Etiology, Risk Factors and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) birth cohort study that included children from eight countries. Methods Non-diarrheal stool samples (N = 22,846) from 1253 children from multiple sites were evaluated for a panel of 40 enteropathogens and fecal concentrations of myeloperoxidase, alpha-1-antitrypsin, and neopterin. Among these same children, urinary lactulose:mannitol (L:M) (N = 6363) and plasma alpha-1-acid glycoprotein (AGP) (N = 2797) were also measured. The temporal sampling design was used to create a directed acyclic graph of proposed mechanistic pathways between enteropathogen detection in non-diarrheal stools, biomarkers of intestinal permeability and inflammation, systemic inflammation and change in length- and weight- for age in children 0–2 years of age. Findings Children in these populations had frequent enteric infections and high levels of both intestinal and systemic inflammation. Higher burdens of enteropathogens, especially those categorized as being enteroinvasive or causing mucosal disruption, were associated with elevated biomarker concentrations of gut and systemic inflammation and, via these associations, indirectly associated with both reduced linear and ponderal growth. Evidence for the association with reduced linear growth was stronger for systemic inflammation than for gut inflammation; the opposite was true of reduced ponderal growth. Although Giardia was associated with reduced growth, the association was not mediated by any of the biomarkers evaluated. Interpretation The large quantity of empirical evidence contributing to this analysis supports the conceptual model of EE. The effects of EE on growth faltering in young children were small, but multiple mechanistic pathways underlying the attribution of growth failure to asymptomatic enteric infections had statistical support in the analysis. The strongest evidence for EE was the association between enteropathogens and linear growth mediated through systemic inflammation. Funding Bill & Melinda Gates Foundation.
Collapse
|
183
|
Ramakrishnan G, Petri WA. Secondary Carnitine Deficiency in Environmental Enteric Dysfunction. EBioMedicine 2017; 17:9-10. [PMID: 28254256 PMCID: PMC5360577 DOI: 10.1016/j.ebiom.2017.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 11/15/2022] Open
Affiliation(s)
- Girija Ramakrishnan
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, USA.
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, USA
| |
Collapse
|
184
|
Wang AZ, Shulman RJ, Crocker AH, Thakwalakwa C, Maleta KM, Devaraj S, Manary MJ, Trehan I. A Combined Intervention of Zinc, Multiple Micronutrients, and Albendazole Does Not Ameliorate Environmental Enteric Dysfunction or Stunting in Rural Malawian Children in a Double-Blind Randomized Controlled Trial. J Nutr 2017; 147:97-103. [PMID: 27807040 DOI: 10.3945/jn.116.237735] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) and linear growth stunting affect many rural agrarian children in the developing world and contribute to the persistently high rates of stunting that are observed worldwide. Effective interventions to consistently ameliorate EED are lacking. OBJECTIVE We tested whether a bundle of safe and affordable interventions would decrease EED and stunting over 12-24 wk in a cohort of rural Malawian children 12-35 mo old. METHODS This was a randomized, double-blind, placebo-controlled clinical trial in which the intervention group received a single dose of albendazole and 14 d of zinc at enrollment and after 20 wk. The intervention group also received a daily multiple micronutrient powder throughout the 24 wk of study. The primary outcomes were improvements in EED, as measured by the urinary lactulose-to-mannitol ratio (L:M ratio) from dual-sugar absorption testing, and linear growth. Urinary L:M ratios and anthropometric measurements were evaluated after 12 and 24 wk of intervention and compared with a placebo group that did not receive any of these interventions. RESULTS A total of 254 children were enrolled at a mean age of 24 mo; 55% were female. Their mean weight-for-age z score was -1.5, and their mean length-for-age z score was -0.9. After 12 and 24 wk of study, increases in the L:M ratio did not differ between the intervention group (0.071 and 0.088 units, respectively) and the placebo group (0.073 and 0.080 units, respectively) (P = 0.87 and 0.19, respectively). Relative changes in length and weight also did not differ significantly between groups at any time point. CONCLUSION The combined usage of albendazole, zinc, and a daily multiple micronutrient powder did not decrease EED or stunting in this population of agrarian children 12-35 mo old in rural Malawi. Alternative interventions to improve these diseases should be investigated. This trial was registered at clinicaltrials.gov as NCT02253095.
Collapse
Affiliation(s)
- Alfred Z Wang
- University of Texas Southwestern Medical School, Dallas, TX.,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Robert J Shulman
- USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,Pediatrics and
| | - Audrey H Crocker
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | | | | | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,School of Public Health and Family Medicine and
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi
| |
Collapse
|
185
|
McCormick BJJ, Lee GO, Seidman JC, Haque R, Mondal D, Quetz J, Lima AAM, Babji S, Kang G, Shrestha SK, Mason CJ, Qureshi S, Bhutta ZA, Olortegui MP, Yori PP, Samie A, Bessong P, Amour C, Mduma E, Patil CL, Guerrant RL, Lang DR, Gottlieb M, Caulfield LE, Kosek MN. Dynamics and Trends in Fecal Biomarkers of Gut Function in Children from 1-24 Months in the MAL-ED Study. Am J Trop Med Hyg 2016; 96:465-472. [PMID: 27994110 PMCID: PMC5303054 DOI: 10.4269/ajtmh.16-0496] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/06/2016] [Indexed: 12/28/2022] Open
Abstract
Growth and development shortfalls that are disproportionately prevalent in children living in poor environmental conditions are postulated to result, at least in part, from abnormal gut function. Using data from The Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) longitudinal cohort study, we examine biomarkers of gut inflammation and permeability in relation to environmental exposures and feeding practices. Trends in the concentrations of three biomarkers, myeloperoxidase (MPO), neopterin (NEO), and α-1-antitrypsin (AAT), are described from fecal samples collected during the first 2 years of each child's life. A total of 22,846 stool samples were processed during the longitudinal sampling of 2,076 children 0–24 months of age. Linear mixed models were constructed to examine the relationship between biomarker concentrations and recent food intake, symptoms of illness, concurrent enteropathogen infection, and socioeconomic status. Average concentrations of MPO, NEO, and AAT were considerably higher than published references for healthy adults. The concentration of each biomarker tended to decrease over the first 2 years of life and was highly variable between samples from each individual child. Both MPO and AAT were significantly elevated by recent breast milk intake. All three biomarkers were associated with pathogen presence, although the strength and direction varied by pathogen. The interpretation of biomarker concentrations is subject to the context of their collection. Herein, we identify that common factors (age, breast milk, and enteric infection) influence the concentration of these biomarkers. Within the context of low- and middle-income communities, we observe concentrations that indicate gut abnormalities, but more appropriate reference standards are needed.
Collapse
Affiliation(s)
| | - Gwenyth O Lee
- Tulane University, New Orleans, Louisiana.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Jessica C Seidman
- Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Dinesh Mondal
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | | | | | | | - Sanjaya K Shrestha
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | - Carl J Mason
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | | | | | - Maribel Paredes Olortegui
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | - Pablo Peñataro Yori
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | | | | | | | | | | | | | - Dennis R Lang
- Foundation for the National Institutes of Health, Bethesda, Maryland.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Michael Gottlieb
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Laura E Caulfield
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Margaret N Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
186
|
Vyas S, Kov P, Smets S, Spears D. Disease externalities and net nutrition: Evidence from changes in sanitation and child height in Cambodia, 2005-2010. ECONOMICS AND HUMAN BIOLOGY 2016; 23:235-245. [PMID: 27776300 PMCID: PMC5147726 DOI: 10.1016/j.ehb.2016.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 05/03/2023]
Abstract
Child height is an important indicator of human capital and human development, in large part because early life health and net nutrition shape both child height and adult economic productivity and health. Between 2005 and 2010, the average height of children under 5 in Cambodia significantly increased. What contributed to this improvement? Recent evidence suggests that exposure to poor sanitation - and specifically to widespread open defecation - can pose a critical threat to child growth. We closely analyze the sanitation height gradient in Cambodia in these two years. Decomposition analysis, in the spirit of Blinder-Oaxaca, suggests that the reduction in children's exposure to open defecation can statistically account for much or all of the increase in average child height between 2005 and 2010. In particular, we see evidence of externalities, indicating an important role for public policy: it is the sanitation behavior of a child's neighbors that matters more for child height rather than the household's sanitation behavior by itself. Moving from an area in which 100% of households defecate in the open to an area in which no households defecate in the open is associated with an average increase in height-for-age z-score of between 0.3 and 0.5. Our estimates are quantitatively robust and comparable with other estimates in the literature.
Collapse
Affiliation(s)
| | - Phyrum Kov
- World Bank Water and Sanitation Program East Asia and the Pacific, 113 Norodom Blvd, Phnom Penh, Cambodia.
| | - Susanna Smets
- World Bank Water and Sanitation Program East Asia and the Pacific, 113 Norodom Blvd, Phnom Penh, Cambodia.
| | - Dean Spears
- Research Institute for Compassionate Economics; Economics and Planning Unit, Indian Statistical Institute, New Delhi, India; University of Texas, Austin, United States.
| |
Collapse
|
187
|
Owino V, Ahmed T, Freemark M, Kelly P, Loy A, Manary M, Loechl C. Environmental Enteric Dysfunction and Growth Failure/Stunting in Global Child Health. Pediatrics 2016; 138:peds.2016-0641. [PMID: 27940670 DOI: 10.1542/peds.2016-0641] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2016] [Indexed: 11/24/2022] Open
Abstract
Approximately 25% of the world's children aged <5 years have stunted growth, which is associated with increased mortality, cognitive dysfunction, and loss of productivity. Reducing by 40% the number of stunted children is a global target for 2030. The pathogenesis of stunting is poorly understood. Prenatal and postnatal nutritional deficits and enteric and systemic infections clearly contribute, but recent findings implicate a central role for environmental enteric dysfunction (EED), a generalized disturbance of small intestinal structure and function found at a high prevalence in children living under unsanitary conditions. Mechanisms contributing to growth failure in EED include intestinal leakiness and heightened permeability, gut inflammation, dysbiosis and bacterial translocation, systemic inflammation, and nutrient malabsorption. Because EED has multiple causal pathways, approaches to manage it need to be multifaceted. Potential interventions to tackle EED include: (1) reduction of exposure to feces and contact with animals through programs such as improved water, sanitation, and hygiene; (2) breastfeeding and enhanced dietary diversity; (3) probiotics and prebiotics; (4) nutrient supplements, including zinc, polyunsaturated fatty acids, and amino acids; (5) antiinflammatory agents such as 5-aminosalicyclic acid; and (6) antibiotics in the context of acute malnutrition and infection. Better understanding of the underlying causes of EED and development of noninvasive, practical, simple, and affordable point-of-care diagnostic tools remain key gaps. "Omics" technologies (genomics, epigenomics, transcriptomics, proteomics, and metabolomics) and stable isotope techniques (eg, 13C breath tests) targeted at children and their intestinal microbiota will enhance our ability to successfully identify, manage, and prevent this disorder.
Collapse
Affiliation(s)
- Victor Owino
- International Atomic Energy Agency, Vienna, Austria;
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Research, Bangladesh, Dhaka, Bangladesh
| | - Michael Freemark
- Division of Pediatric Endocrinology, Duke University Medical Center, Durham, North Carolina
| | - Paul Kelly
- University of Zambia, Lusaka, Zambia.,Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Alexander Loy
- Department of Microbiology and Ecosystem Science, Research Network "Chemistry meets Microbiology," University of Vienna, Vienna, Austria; and
| | | | | |
Collapse
|
188
|
Uddin MI, Islam S, Nishat NS, Hossain M, Rafique TA, Rashu R, Hoq MR, Zhang Y, Saha A, Harris JB, Calderwood SB, Bhuiyan TR, Ryan ET, Leung DT, Qadri F. Biomarkers of Environmental Enteropathy are Positively Associated with Immune Responses to an Oral Cholera Vaccine in Bangladeshi Children. PLoS Negl Trop Dis 2016; 10:e0005039. [PMID: 27824883 PMCID: PMC5100882 DOI: 10.1371/journal.pntd.0005039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022] Open
Abstract
Environmental enteropathy (EE) is a poorly understood condition that refers to chronic alterations in intestinal permeability, absorption, and inflammation, which mainly affects young children in resource-limited settings. Recently, EE has been linked to suboptimal oral vaccine responses in children, although immunological mechanisms are poorly defined. The objective of this study was to determine host factors associated with immune responses to an oral cholera vaccine (OCV). We measured antibody and memory T cell immune responses to cholera antigens, micronutrient markers in blood, and EE markers in blood and stool from 40 Bangladeshi children aged 3-14 years who received two doses of OCV given 14 days apart. EE markers included stool myeloperoxidase (MPO) and alpha anti-trypsin (AAT), and plasma endotoxin core antibody (EndoCab), intestinal fatty acid binding protein (i-FABP), and soluble CD14 (sCD14). We used multiple linear regression analysis with LASSO regularization to identify host factors, including EE markers, micronutrient (nutritional) status, age, and HAZ score, predictive for each response of interest. We found stool MPO to be positively associated with IgG antibody responses to the B subunit of cholera toxin (P = 0.03) and IgA responses to LPS (P = 0.02); plasma sCD14 to be positively associated with LPS IgG responses (P = 0.07); plasma i-FABP to be positively associated with LPS IgG responses (P = 0.01) and with memory T cell responses specific to cholera toxin (P = 0.01); stool AAT to be negatively associated with IL-10 (regulatory) T cell responses specific to cholera toxin (P = 0.02), and plasma EndoCab to be negatively associated with cholera toxin-specific memory T cell responses (P = 0.02). In summary, in a cohort of children 3-14 years old, we demonstrated that the majority of biomarkers of environmental enteropathy were positively associated with immune responses after vaccination with an OCV.
Collapse
Affiliation(s)
- Muhammad Ikhtear Uddin
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Shahidul Islam
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Naoshin S. Nishat
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Motaher Hossain
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Tanzeem Ahmed Rafique
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Rasheduzzaman Rashu
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Mohammad Rubel Hoq
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Yue Zhang
- Department of Internal Medicine, Division of Epidemiology, University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Amit Saha
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Jason B. Harris
- Department of Immunology and Infectious Diseases, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephen B. Calderwood
- Department of Immunology and Infectious Diseases, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Taufiqur Rahman Bhuiyan
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Edward T. Ryan
- Department of Immunology and Infectious Diseases, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel T. Leung
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- Department of Internal Medicine, Division of Infectious Diseases, University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Firdausi Qadri
- Mucosal Immunology and Vaccinology Laboratory, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- * E-mail:
| |
Collapse
|
189
|
Twitchell EL, Tin C, Wen K, Zhang H, Becker-Dreps S, Azcarate-Peril MA, Vilchez S, Li G, Ramesh A, Weiss M, Lei S, Bui T, Yang X, Schultz-Cherry S, Yuan L. Modeling human enteric dysbiosis and rotavirus immunity in gnotobiotic pigs. Gut Pathog 2016; 8:51. [PMID: 27826359 PMCID: PMC5100090 DOI: 10.1186/s13099-016-0136-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/20/2016] [Indexed: 01/19/2023] Open
Abstract
Background Rotavirus vaccines have poor efficacy in infants from low- and middle-income countries. Gut microbiota is thought to influence the immune response to oral vaccines. Thus, we developed a gnotobiotic (Gn) pig model of enteric dysbiosis to study the effects of human gut microbiota (HGM) on immune responses to rotavirus vaccination, and the effects of rotavirus challenge on the HGM by colonizing Gn pigs with healthy HGM (HHGM) or unhealthy HGM (UHGM). The UHGM was from a Nicaraguan infant with a high enteropathy score (ES) and no seroconversion following administration of oral rotavirus vaccine, while the converse was characteristic of the HHGM. Pigs were vaccinated, a subset was challenged, and immune responses and gut microbiota were evaluated. Results Significantly more rotavirus-specific IFN-γ producing T cells were in the ileum, spleen, and blood of HHGM than those in UHGM pigs after three vaccine doses, suggesting HHGM induces stronger cell-mediated immunity than UHGM. There were significant correlations between multiple Operational Taxonomic Units (OTUs) and frequencies of IFN-γ producing T cells at the time of challenge. There were significant positive correlations between Collinsella and CD8+ T cells in blood and ileum, as well as CD4+ T cells in blood, whereas significant negative correlations between Clostridium and Anaerococcus, and ileal CD8+ and CD4+ T cells. Differences in alpha diversity and relative abundances of OTUs were detected between the groups both before and after rotavirus challenge. Conclusion Alterations in microbiome diversity and composition along with correlations between certain microbial taxa and T cell responses warrant further investigation into the role of the gut microbiota and certain microbial species on enteric immunity. Our results support the use of HGM transplanted Gn pigs as a model of human dysbiosis during enteric infection, and oral vaccine responses. Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0136-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erica L Twitchell
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Christine Tin
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Husen Zhang
- Microbiome Core, Cancer Inflammation Program, National Cancer Institute, Bethesda, MD USA
| | - Sylvia Becker-Dreps
- Department of Family Medicine, University of North Carolina School of Medicine, Chapel Hill, NC USA
| | - M Andrea Azcarate-Peril
- Department of Cell Biology and Physiology, School of Medicine and Microbiome Core Facility, Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC USA
| | - Samuel Vilchez
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León, Nicaragua
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Ashwin Ramesh
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Mariah Weiss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Shaohua Lei
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Tammy Bui
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| |
Collapse
|
190
|
Blackwell AD, Urlacher SS, Beheim B, von Rueden C, Jaeggi A, Stieglitz J, Trumble BC, Gurven M, Kaplan H. Growth references for Tsimane forager-horticulturalists of the Bolivian Amazon. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2016; 162:441-461. [PMID: 28218400 DOI: 10.1002/ajpa.23128] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 11/10/2022]
Abstract
OBJECTIVES Growth standards and references currently used to assess population and individual health are derived primarily from urban populations, including few individuals from indigenous or subsistence groups. Given environmental and genetic differences, growth may vary in these populations. Thus, there is a need to assess whether international standards are appropriate for all populations, and to produce population specific references if growth differs. Here we present and assess growth references for the Tsimane, an indigenous population of Bolivian forager-horticulturalists. METHODS Mixed cross-sectional/longitudinal anthropometrics (9,614 individuals; 30,118 observations; ages 0-29 years) were used to generate centile curves and Lambda-Mu-Sigma (LMS) tables for height-for-age, weight-for-age, body mass index (BMI)-for-age, and weight-for-height (WFH) using Generalized Additive Models for Location Shape and Scale (GAMLSS). Velocity curves were generated using SuperImposition by Translation and Rotation (SITAR). Tsimane ≤5 years were compared to World Health Organization (WHO) standards while those >5 years were compared to WHO school age references. All ages were compared to published references for Shuar forager-horticulturalists of the Ecuadorian Amazon. RESULTS Tsimane growth differs from WHO values in height and weight, but is similar for BMI and WFH. Tsimane growth is characterized by slow height velocity in childhood and early adolescent peak height velocity at 11.3 and 13.2 years for girls and boys. Tsimane growth patterns are similar to Shuar, suggesting shared features of growth among indigenous South Americans. CONCLUSIONS International references for BMI-for-age and WFH are likely appropriate for Tsimane, but differences in height-for-age and weight-for-age suggest Tsimane specific references may be useful for these measures.
Collapse
Affiliation(s)
- Aaron D Blackwell
- Department of Anthropology, University of California Santa Barbara, California.,Tsimane Health and Life History Project, San Borja, Bolivia
| | - Samuel S Urlacher
- Department of Anthropology, Hunter College, City University of New York, New York
| | - Bret Beheim
- Tsimane Health and Life History Project, San Borja, Bolivia.,Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.,Department of Anthropology, University of New Mexico, Albuquerque, New Mexico
| | - Christopher von Rueden
- Tsimane Health and Life History Project, San Borja, Bolivia.,Jepson School of Leadership Studies, University of Richmond, Richmond, Virginia
| | - Adrian Jaeggi
- Tsimane Health and Life History Project, San Borja, Bolivia.,Department of Anthropology, Emory University, Atlanta, Georgia
| | - Jonathan Stieglitz
- Tsimane Health and Life History Project, San Borja, Bolivia.,Department of Anthropology, University of New Mexico, Albuquerque, New Mexico.,Institute for Advanced Study in Toulouse, Toulouse, France
| | - Benjamin C Trumble
- Department of Anthropology, University of California Santa Barbara, California.,Tsimane Health and Life History Project, San Borja, Bolivia.,Center for Evolution and Medicine, Arizona State University, Tempe, Arizona.,School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona
| | - Michael Gurven
- Department of Anthropology, University of California Santa Barbara, California.,Tsimane Health and Life History Project, San Borja, Bolivia
| | - Hillard Kaplan
- Tsimane Health and Life History Project, San Borja, Bolivia.,Department of Anthropology, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
191
|
Perin J, Thomas A, Oldja L, Ahmed S, Parvin T, Bhuyian SI, Sarker B, Biswas SK, Faruque ASG, Sack RB, George CM. Geophagy Is Associated with Growth Faltering in Children in Rural Bangladesh. J Pediatr 2016; 178:34-39.e1. [PMID: 27496267 DOI: 10.1016/j.jpeds.2016.06.077] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/10/2016] [Accepted: 06/27/2016] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To determine the relationship between geophagy (mouthing of dirt, sand, clay, or mud) and growth faltering in young children. STUDY DESIGN We examined linear growth as height and weight standardized by age and sex, and weight standardized by height, in a cohort of children aged 6-36 months in rural Mirzapur, Bangladesh. We determined geophagy behavior at baseline through caregiver report. Anthropometric measurements were assessed at baseline and at a 1-year follow-up. RESULTS We found that among children not stunted at baseline, those with caregiver-reported geophagy at baseline grew less over 1 year compared with their peers, with a difference in the change of standardized height for age and sex of -0.31 (95% CI, -0.61 to -0.01). CONCLUSION These findings show that caregiver-reported geophagy was associated with growth faltering in a pediatric population in rural Bangladesh. Future studies are needed to learn more about this exposure pathway and its relevance to child growth.
Collapse
Affiliation(s)
- Jamie Perin
- International Health, Johns Hopkins University, Baltimore, MD.
| | - Alvin Thomas
- International Health, Johns Hopkins University, Baltimore, MD
| | - Lauren Oldja
- International Health, Johns Hopkins University, Baltimore, MD
| | - Shahnawaz Ahmed
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmina Parvin
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Bidduth Sarker
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shwapon K Biswas
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh; Department of Internal Medicine, Rangpur Medical College Hospital, Rangpur, Bangladesh
| | - Abu S G Faruque
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - R Bradley Sack
- International Health, Johns Hopkins University, Baltimore, MD
| | | |
Collapse
|
192
|
Environmental Enteric Dysfunction Is Associated With Poor Linear Growth and Can Be Identified by Host Fecal mRNAs. J Pediatr Gastroenterol Nutr 2016; 63:453-459. [PMID: 27347722 PMCID: PMC5084633 DOI: 10.1097/mpg.0000000000001315] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Environmental enteric dysfunction (EED) can be assessed by the lactulose:mannitol (L:M) test. Our objective was to determine if selected host fecal transcripts were correlated with EED, and whether transcripts and clinical characteristics could be used to predict EED in rural African children. METHODS Demographic and sanitation characteristics, along with L:M testing and host fecal transcript analyses from 798 asymptomatic Malawian children aged 12 to 61 months were compared with linear growth over the subsequent 3 months. Fecal host mRNA analysis included quantification of expression of 18 transcripts associated with L:M. Permeability was categorized as normal (L:M ≤ 0.15), moderate (0.15<L:M<0.45) and severe (L:M ≥ 0.45), and random forest predictive models were created. RESULTS L:M was inversely correlated with linear growth over the subsequent 3 months (r = -0.32, P < 0.001) and severe EED was associated with stunting (P < 0.0001). Age younger than 24 months, weight-for-height z score <0, domesticated animals in the child's sleep environment, lack of a pit latrine combined with a potentially contaminated water source, and a recent history of diarrhea were associated with severe EED. A random forest model using CD53, HLA-DRA, MUC12, and TNF was 84% sensitive for severe EED and 83% sensitive for no EED. CONCLUSIONS Selected host fecal transcripts can be used in a random forest model as a noninvasive biomarker for categories of EED in rural African children.
Collapse
|
193
|
Chronic Health Consequences of Acute Enteric Infections in the Developing World. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
194
|
Guerrant RL, Leite AM, Pinkerton R, Medeiros PHQS, Cavalcante PA, DeBoer M, Kosek M, Duggan C, Gewirtz A, Kagan JC, Gauthier AE, Swann J, Mayneris-Perxachs J, Bolick DT, Maier EA, Guedes MM, Moore SR, Petri WA, Havt A, Lima IF, Prata MDMG, Michaleckyj JC, Scharf RJ, Sturgeon C, Fasano A, Lima AAM. Biomarkers of Environmental Enteropathy, Inflammation, Stunting, and Impaired Growth in Children in Northeast Brazil. PLoS One 2016; 11:e0158772. [PMID: 27690129 PMCID: PMC5045163 DOI: 10.1371/journal.pone.0158772] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/21/2016] [Indexed: 01/27/2023] Open
Abstract
Critical to the design and assessment of interventions for enteropathy and its developmental consequences in children living in impoverished conditions are non-invasive biomarkers that can detect intestinal damage and predict its effects on growth and development. We therefore assessed fecal, urinary and systemic biomarkers of enteropathy and growth predictors in 375 6–26 month-old children with varying degrees of malnutrition (stunting or wasting) in Northeast Brazil. 301 of these children returned for followup anthropometry after 2-6m. Biomarkers that correlated with stunting included plasma IgA anti-LPS and anti-FliC, zonulin (if >12m old), and intestinal FABP (I-FABP, suggesting prior barrier disruption); and with citrulline, tryptophan and with lower serum amyloid A (SAA) (suggesting impaired defenses). In contrast, subsequent growth was predicted in those with higher fecal MPO or A1AT and also by higher L/M, plasma LPS, I-FABP and SAA (showing intestinal barrier disruption and inflammation). Better growth was predicted in girls with higher plasma citrulline and in boys with higher plasma tryptophan. Interactions were also seen with fecal MPO and neopterin in predicting subsequent growth impairment. Biomarkers clustered into markers of 1) functional intestinal barrier disruption and translocation, 2) structural intestinal barrier disruption and inflammation and 3) systemic inflammation. Principle components pathway analyses also showed that L/M with %L, I-FABP and MPO associate with impaired growth, while also (like MPO) associating with a systemic inflammation cluster of kynurenine, LBP, sCD14, SAA and K/T. Systemic evidence of LPS translocation associated with stunting, while markers of barrier disruption or repair (A1AT and Reg1 with low zonulin) associated with fecal MPO and neopterin. We conclude that key noninvasive biomarkers of intestinal barrier disruption, LPS translocation and of intestinal and systemic inflammation can help elucidate how we recognize, understand, and assess effective interventions for enteropathy and its growth and developmental consequences in children in impoverished settings.
Collapse
Affiliation(s)
- Richard L. Guerrant
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
- * E-mail:
| | - Alvaro M. Leite
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | - Relana Pinkerton
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | | | | | - Mark DeBoer
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Margaret Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Christopher Duggan
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | - Andrew Gewirtz
- Institute for Biomedical Sciences in the Center for Inflammation, Immunity and Infection at Georgia State University, Atlanta, GA, United States of America
| | - Jonathan C. Kagan
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | - Anna E. Gauthier
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | | | | | - David T. Bolick
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Elizabeth A. Maier
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - Marjorie M. Guedes
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - Sean R. Moore
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - William A. Petri
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Alexandre Havt
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | - Ila F. Lima
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | | | - Josyf C. Michaleckyj
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Rebecca J. Scharf
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Craig Sturgeon
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition at Massachusetts General Hospital for Children, Harvard University, Boston, MA, United States of America
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition at Massachusetts General Hospital for Children, Harvard University, Boston, MA, United States of America
| | - Aldo A. M. Lima
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| |
Collapse
|
195
|
Gough EK, Prendergast AJ, Mutasa KE, Stoltzfus RJ, Manges AR. Assessing the Intestinal Microbiota in the SHINE Trial. Clin Infect Dis 2016; 61 Suppl 7:S738-44. [PMID: 26602302 PMCID: PMC4657595 DOI: 10.1093/cid/civ850] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Advances in DNA sequencing technology now allow us to explore the dynamics and functions of the microbes that inhabit the human body, the microbiota. Recent studies involving experimental animal models suggest a role of the gut microbiota in growth. However, the specific changes in the human gut microbiota that contribute to growth remain unclear, and studies investigating the gut microbiota as a determinant of environmental enteric dysfunction (EED) and child stunting are lacking. In this article, we review the evidence for a link between the developing infant gut microbiota, infant feeding, EED, and stunting, and discuss the potential causal pathways relating these variables. We outline the analytic approaches we will use to investigate these relationships, by capitalizing on the longitudinal design and randomized interventions of the Sanitation Hygiene Infant Nutrition Efficacy trial in Zimbabwe.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe Blizard Institute, Queen Mary University of London, United Kingdom
| | - Kuda E Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
196
|
Prendergast AJ, Humphrey JH, Mutasa K, Majo FD, Rukobo S, Govha M, Mbuya MNN, Moulton LH, Stoltzfus RJ. Assessment of Environmental Enteric Dysfunction in the SHINE Trial: Methods and Challenges. Clin Infect Dis 2016; 61 Suppl 7:S726-32. [PMID: 26602300 PMCID: PMC4657593 DOI: 10.1093/cid/civ848] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Environmental enteric dysfunction (EED) is a virtually ubiquitous, but poorly defined, disorder of the small intestine among people living in conditions of poverty, which begins early in infancy and persists. EED is characterized by altered gut structure and function, leading to reduced absorptive surface area and impaired intestinal barrier function. It is hypothesized that recurrent exposure to fecal pathogens and changes in the composition of the intestinal microbiota initiate this process, which leads to a self-perpetuating cycle of pathology. We view EED as a primary gut disorder that drives chronic systemic inflammation, leading to growth hormone resistance and impaired linear growth. There is currently no accepted case definition or gold-standard biomarker of EED, making field studies challenging. The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) trial in Zimbabwe is evaluating the independent and combined effects of a package of infant feeding and/or water, sanitation, and hygiene interventions on stunting and anemia. SHINE therefore provides an opportunity to longitudinally evaluate EED in a well-characterized cohort of infants, using a panel of biomarkers along the hypothesized causal pathway. Our aims are to describe the evolution of EED during infancy, ascertain its contribution to stunting, and investigate the impact of the randomized interventions on the EED pathway. In this article, we describe current concepts of EED, challenges in defining the condition, and our approach to evaluating EED in the SHINE trial.
Collapse
Affiliation(s)
- Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe Blizard Institute, Queen Mary University of London, United Kingdom Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mduduzi N N Mbuya
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Lawrence H Moulton
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | | |
Collapse
|
197
|
Humphrey JH, Jones AD, Manges A, Mangwadu G, Maluccio JA, Mbuya MNN, Moulton LH, Ntozini R, Prendergast AJ, Stoltzfus RJ, Tielsch JM. The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) Trial: Rationale, Design, and Methods. Clin Infect Dis 2016; 61 Suppl 7:S685-702. [PMID: 26602296 PMCID: PMC4657589 DOI: 10.1093/cid/civ844] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Child stunting and anemia are intractable public health problems in developing countries and have profound short- and long-term consequences. The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) trial is motivated by the premise that environmental enteric dysfunction (EED) is a major underlying cause of both stunting and anemia, that chronic inflammation is the central characteristic of EED mediating these adverse effects, and that EED is primarily caused by high fecal ingestion due to living in conditions of poor water, sanitation, and hygiene (WASH). SHINE is a proof-of-concept, 2 × 2 factorial, cluster-randomized, community-based trial in 2 rural districts of Zimbabwe that will test the independent and combined effects of protecting babies from fecal ingestion (factor 1, operationalized through a WASH intervention) and optimizing nutritional adequacy of infant diet (factor 2, operationalized through an infant and young child feeding [IYCF] intervention) on length and hemoglobin at 18 months of age. Within SHINE we will measure 2 causal pathways. The program impact pathway comprises the series of processes and behaviors linking implementation of the interventions with the 2 child health primary outcomes; it will be modeled using measures of fidelity of intervention delivery and household uptake of promoted behaviors and practices. We will also measure a range of household and individual characteristics, social interactions, and maternal capabilities for childcare, which we hypothesize will explain heterogeneity along these pathways. The biomedical pathway comprises the infant biologic responses to the WASH and IYCF interventions that ultimately result in attained stature and hemoglobin concentration at 18 months of age; it will be elucidated by measuring biomarkers of intestinal structure and function (inflammation, regeneration, absorption, and permeability); microbial translocation; systemic inflammation; and hormonal determinants of growth and anemia among a subgroup of infants enrolled in an EED substudy. This article describes the rationale, design, and methods underlying the SHINE trial. CLINICAL TRIALS REGISTRATION NCT01824940.
Collapse
|
198
|
Morita T, Godfrey S, George CM. Systematic review of evidence on the effectiveness of safe child faeces disposal interventions. Trop Med Int Health 2016; 21:1403-1419. [PMID: 27546207 DOI: 10.1111/tmi.12773] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To review and synthesise the available evidence on the effectiveness of interventions targeting unsafe child faeces disposal in reducing this behaviour and improving child health in low- and middle-income countries. METHODS PubMed and EMBASE were systematically searched. Studies meeting the inclusion criteria were reviewed and key information on study methodologies and outcomes were extracted. RESULTS A total of 1048 articles were screened, and eight studies representing five countries were included for the review. Three were randomised controlled trials, and five were prospective cohort studies. There was wide variability across studies in the definition of 'safe disposal' of child faeces. Six studies reported the change in child faeces disposal practices associated with safe child faeces disposal interventions. However, only one study found a significant improvement in this behaviour. Two of the six studies that evaluated the health impact of delivered interventions found significant reductions in childhood diarrhoea associated with safe faeces disposal practices, and one study reported a positive effect on child growth and ascariasis. Only one study was identified that delivered a single intervention solely focused on safe child faeces disposal. Unfortunately, this study did not investigate the impact of this intervention on child health. CONCLUSIONS There are major methodological limitations in studies that assessed the impact of safe child faeces disposal interventions. The health impact of these interventions is inconclusive because the quality of the current evidence is poor. Randomised controlled trials are urgently needed to assess the impact of safe faeces disposal interventions on child health.
Collapse
Affiliation(s)
- Tomohiko Morita
- Program in Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Samuel Godfrey
- Water, Sanitation and Hygiene Section, UNICEF Ethiopia, Addis Ababa, Ethiopia
| | - Christine Marie George
- Program in Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
199
|
George CM, Oldja L, Biswas S, Perin J, Sack RB, Ahmed S, Shahnaij M, Haque R, Parvin T, Azmi IJ, Bhuyian SI, Talukder KA, Faruque AG. Unsafe Child Feces Disposal is Associated with Environmental Enteropathy and Impaired Growth. J Pediatr 2016; 176:43-9. [PMID: 27318380 DOI: 10.1016/j.jpeds.2016.05.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/28/2016] [Accepted: 05/11/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To investigate the relationship between unsafe child feces disposal, environmental enteropathy, and impaired growth, we conducted a prospective cohort study of 216 young children in rural Bangladesh. STUDY DESIGN Using a prospective cohort study design in rural Bangladesh, unsafe child feces disposal, using the Joint Monitoring Program definition, was assessed using 5-hour structured observation by trained study personnel as well as caregiver reports. Anthropometric measurements were collected at baseline and at a 9-month follow-up. Stool was analyzed for fecal markers of environmental enteropathy: alpha-1-antitrypsin, myeloperoxidase, neopterin (combined to form an environmental enteropathy disease activity score), and calprotectin. FINDINGS Among 216 households with young children, 84% had an unsafe child feces disposal event during structured observation and 75% had caregiver reported events. There was no significant difference in observed unsafe child feces disposal events for households with or without an improved sanitation option (82% vs 85%, P = .72) or by child's age (P = .96). Children in households where caregivers reported unsafe child feces disposal had significantly higher environmental enteropathy scores (0.82-point difference, 95% CI 0.11-1.53), and significantly greater odds of being wasted (weight-for-height z score <-2 SDs) (9% vs 0%, P = .024). In addition, children in households with observed unsafe feces disposal had significantly reduced change in weight-for-age z-score (-0.34 [95% CI -0.68, -0.01] and weight-for-height z score (-0.52 [95% CI -0.98, -0.06]). CONCLUSION Unsafe child feces disposal was significantly associated with environmental enteropathy and impaired growth in a pediatric population in rural Bangladesh. Interventions are needed to reduce this high-risk behavior to protect the health of susceptible pediatric populations.
Collapse
Affiliation(s)
- Christine Marie George
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Lauren Oldja
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Shwapon Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Jamie Perin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - R Bradley Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Shahnawaz Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Mohammad Shahnaij
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Ishrat J Azmi
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Sazzadul Islam Bhuyian
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Kaisar A Talukder
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| | - Abu G Faruque
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) Dhaka, Bangladesh
| |
Collapse
|
200
|
Burke RM, Suchdev PS, Rebolledo PA, de Aceituno AMF, Revollo R, Iñiguez V, Klein M, Drews-Botsch C, Leon JS. Predictors of Inflammation in a Cohort of Bolivian Infants and Toddlers. Am J Trop Med Hyg 2016; 95:954-963. [PMID: 27527627 DOI: 10.4269/ajtmh.16-0292] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/06/2016] [Indexed: 12/28/2022] Open
Abstract
Inflammation has been associated with cardiovascular disease and other health outcomes in children and adults, yet few longitudinal data are available on prevalence and predictors of inflammation in infants. We aimed to identify the prevalence of inflammation in a cohort of Bolivian infants and estimate its association with acute (recent illnesses) and chronic (overweight, stunting) morbidities and potential pathogen exposure (represented by water, sanitation, and hygiene [WASH] resources). We measured plasma concentrations of two acute phase proteins (C-reactive protein [CRP], marking acute inflammation, and alpha(1)-acid-glycoprotein [AGP], marking chronic inflammation) at three time points (target 2, 6-8, and 12-18 months). Of 451 singleton infants enrolled in the parent study, 272 had the first blood draw and complete data. Anthropometry and sociodemographic and recent illness data (2-week recall of cough, diarrhea, and fever) were collected at each visit. Inflammation was defined as CRP > 5 mg/L or AGP > 1 g/L. The prevalence of inflammation increased from early infancy (3% at first blood draw) to later infancy (15-22% at later blood draws). Recent cough, recent fever, and age in months were significantly associated with relative increases in CRP (7-44%) and AGP (5-23%), whereas recent diarrhea was only significantly associated with an increase in CRP (48%). Neither anthropometry nor WASH was significantly associated with inflammation. Results confirm the role of recent acute illness in inflammation in infants, and indicate that adiposity and WASH are not as important to inflammation in this age category.
Collapse
Affiliation(s)
- Rachel M Burke
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.
| | - Parminder S Suchdev
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia. Emory School of Medicine, Atlanta, Georgia. Nutrition Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Paulina A Rebolledo
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia. Emory School of Medicine, Atlanta, Georgia
| | | | - Rita Revollo
- Servicio Departamental de Salud, La Paz, Bolivia
| | - Volga Iñiguez
- Instituto de Biología Molecular y Biotecnología, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - Mitchel Klein
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Carolyn Drews-Botsch
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Juan S Leon
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| |
Collapse
|